Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Prog Neurobiol ; 216: 102313, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35760142

RESUMEN

We have uncovered a novel role for astrocytes-derived extracellular vesicles (EVs) in controlling intraneuronal Ca2+ concentration ([Ca2+]i) and identified transglutaminase-2 (TG2) as a surface-cargo of astrocytes-derived EVs. Incubation of hippocampal neurons with primed astrocyte-derived EVs have led to an increase in [Ca2+]i, unlike EVs from TG2-knockout astrocytes. Exposure of neurons or brain slices to extracellular TG2 promoted a [Ca2+]i rise, which was reversible upon TG2 removal and was dependent on Ca2+ influx through the plasma membrane. Patch-clamp and calcium imaging recordings revealed TG2-dependent neuronal membrane depolarization and activation of inward currents, due to the Na+/Ca2+-exchanger (NCX) operating in the reverse mode and indirect activation of L-type VOCCs, as indicated by VOCCs/NCX pharmacological inhibitors. A subunit of Na+/K+-ATPase was selected by comparative proteomics and identified as being functionally inhibited by extracellular TG2, implicating Na+/K+-ATPase inhibition in NCX reverse mode-switching leading to Ca2+ influx and higher basal [Ca2+]i. These data suggest that reactive astrocytes control intraneuronal [Ca2+]i through release of EVs with TG2 as responsible cargo, which could have a significant impact on synaptic activity in brain inflammation.


Asunto(s)
Astrocitos , Vesículas Extracelulares , Adenosina Trifosfatasas , Astrocitos/metabolismo , Calcio/metabolismo , Vesículas Extracelulares/metabolismo , Homeostasis , Humanos , Neuronas/metabolismo , Proteína Glutamina Gamma Glutamiltransferasa 2 , Intercambiador de Sodio-Calcio/metabolismo
2.
Exp Neurol ; 342: 113727, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33930392

RESUMEN

Specific neurophysiological seizure patterns in patients with focal epilepsy depend on cerebral location and the underlying neuropathology. Location-specific patterns have been also reported in experimental models. Two focal seizure patterns, named p-type and l-type, typical of neocortical and mesial temporal regions were identified in both patients explored with intracerebral EEG and in animal models. These two patterns were recorded in the olfactory regions and in the entorhinal cortex after either 4AP or BMI administration. Here we mapped epileptiform activities in other cortices to verify the existence of specific epileptiform patterns. Field potentials were simultaneously recorded at multiple locations in olfactory, limbic and neocortical regions of the isolated guinea pig brain after arterial administration of either 4AP or BMI. Most neocortical areas did not generate new distinctive focal seizure-like event (SLE), beside the p-type and l-type patterns. Spiking activity was typically recorded after BMI in all new analyzed regions, whereas SLEs were commonly observed during 4AP perfusion. We confirmed the presence of reproducible region-specific epileptiform patterns in all explored cortical areas and demonstrated that strongly inter-connected areas generate similar SLEs. Our study suggests that p- and l-type SLE represent the most common focal seizure patterns during acute manipulations with pro-epileptic compounds.


Asunto(s)
Mapeo Encefálico/métodos , Encéfalo/fisiopatología , Convulsiones/fisiopatología , Animales , Bicuculina/toxicidad , Encéfalo/efectos de los fármacos , Electroencefalografía/métodos , Femenino , Cobayas , Técnicas de Cultivo de Órganos , Convulsiones/inducido químicamente
3.
Mol Neurobiol ; 58(6): 2824-2835, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33511502

RESUMEN

In light of previous results, we assessed whether liposomes functionalized with ApoE-derived peptide (mApoE) and phosphatidic acid (PA) (mApoE-PA-LIP) impacted on intracellular calcium (Ca2+) dynamics in cultured human cerebral microvascular endothelial cells (hCMEC/D3), as an in vitro human blood-brain barrier (BBB) model, and in cultured astrocytes. mApoE-PA-LIP pre-treatment actively increased both the duration and the area under the curve (A.U.C) of the ATP-evoked Ca2+ waves in cultured hCMEC/D3 cells as well as in cultured astrocytes. mApoE-PA-LIP increased the ATP-evoked intracellular Ca2+ waves even under 0 [Ca2+]e conditions, thus indicating that the increased intracellular Ca2+ response to ATP is mainly due to endogenous Ca2+ release. Indeed, when Sarco-Endoplasmic Reticulum Calcium ATPase (SERCA) activity was blocked by cyclopiazonic acid (CPA), the extracellular application of ATP failed to trigger any intracellular Ca2+ waves, indicating that metabotropic purinergic receptors (P2Y) are mainly involved in the mApoE-PA-LIP-induced increase of the Ca2+ wave triggered by ATP. In conclusion, mApoE-PA-LIP modulate intracellular Ca2+ dynamics evoked by ATP when SERCA is active through inositol-1,4,5-trisphosphate-dependent (InsP3) endoplasmic reticulum Ca2+ release. Considering that P2Y receptors represent important pharmacological targets to treat cognitive dysfunctions, and that P2Y receptors have neuroprotective effects in neuroinflammatory processes, the enhancement of purinergic signaling provided by mApoE-PA-LIP could counteract Aß-induced vasoconstriction and reduction in cerebral blood flow (CBF). Our obtained results could give an additional support to promote mApoE-PA-LIP as effective therapeutic tool for Alzheimer's disease (AD).


Asunto(s)
Enfermedad de Alzheimer/patología , Astrocitos/metabolismo , Encéfalo/patología , Señalización del Calcio , Células Endoteliales/metabolismo , Microvasos/patología , Receptores Purinérgicos/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Astrocitos/efectos de los fármacos , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Línea Celular , Retículo Endoplásmico/metabolismo , Células Endoteliales/efectos de los fármacos , Humanos , Indoles/farmacología , Liposomas , Ácidos Fosfatidicos/química , Ratas , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo
4.
Cell Mol Life Sci ; 77(11): 2235-2253, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31473770

RESUMEN

Neurovascular coupling (NVC) is the mechanism whereby an increase in neuronal activity causes an increase in local cerebral blood flow (CBF) to ensure local supply of oxygen and nutrients to the activated areas. The excitatory neurotransmitter glutamate gates post-synaptic N-methyl-D-aspartate receptors to mediate extracellular Ca2+ entry and stimulate neuronal nitric oxide (NO) synthase to release NO, thereby triggering NVC. Recent work suggested that endothelial Ca2+ signals could underpin NVC by recruiting the endothelial NO synthase. For instance, acetylcholine induced intracellular Ca2+ signals followed by NO release by activating muscarinic 5 receptors in hCMEC/D3 cells, a widely employed model of human brain microvascular endothelial cells. Herein, we sought to assess whether also glutamate elicits metabotropic Ca2+ signals and NO release in hCMEC/D3 cells. Glutamate induced a dose-dependent increase in intracellular Ca2+ concentration ([Ca2+]i) that was blocked by α-methyl-4-carboxyphenylglycine and phenocopied by trans-1-amino-1,3-cyclopentanedicarboxylic acid, which, respectively, block and activate group 1 metabotropic glutamate receptors (mGluRs). Accordingly, hCMEC/D3 expressed both mGluR1 and mGluR5 and the Ca2+ response to glutamate was inhibited by their pharmacological blockade with, respectively, CPCCOEt and MTEP hydrochloride. The Ca2+ response to glutamate was initiated by endogenous Ca2+ release from the endoplasmic reticulum and endolysosomal Ca2+ store through inositol-1,4,5-trisphosphate receptors and two-pore channels, respectively, and sustained by store-operated Ca2+ entry. In addition, glutamate induced robust NO release that was suppressed by pharmacological blockade of the accompanying increase in [Ca2+]i. These data demonstrate for the first time that glutamate may induce metabotropic Ca2+ signals in human brain microvascular endothelial cells. The Ca2+ response to glutamate is likely to support NVC during neuronal activity, thereby reinforcing the emerging role of brain microvascular endothelial cells in the regulation of CBF.


Asunto(s)
Encéfalo/irrigación sanguínea , Señalización del Calcio , Células Endoteliales/metabolismo , Ácido Glutámico/metabolismo , Acoplamiento Neurovascular , Receptores de Glutamato Metabotrópico/metabolismo , Línea Celular , Células Endoteliales/citología , Humanos , Microvasos/citología , Microvasos/metabolismo , Óxido Nítrico/metabolismo
5.
J Cell Physiol ; 235(2): 1515-1530, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31310018

RESUMEN

The neuromodulator histamine is able to vasorelax in human cerebral, meningeal and temporal arteries via endothelial histamine 1 receptors (H1 Rs) which result in the downstream production of nitric oxide (NO), the most powerful vasodilator transmitter in the brain. Although endothelial Ca 2+ signals drive histamine-induced NO release throughout the peripheral circulation, the mechanism by which histamine evokes NO production in human cerebrovascular endothelial cells is still unknown. Herein, we exploited the human cerebral microvascular endothelial cell line, hCMEC/D3, to assess the role of intracellular Ca 2+ signaling in histamine-induced NO release. To achieve this goal, hCMEC/D3 cells were loaded with the Ca 2+ - and NO-sensitive dyes, Fura-2/AM and DAF-FM/AM, respectively. Histamine elicited repetitive oscillations in intracellular Ca 2+ concentration in hCMEC/D3 cells throughout a concentration range spanning from 1 pM up to 300 µM. The oscillatory Ca 2+ response was suppressed by the inhibition of H 1 Rs with pyrilamine, whereas H 1 R was abundantly expressed at the protein level. We further found that histamine-induced intracellular Ca 2+ oscillations were initiated by endogenous Ca 2+ mobilization through inositol-1,4,5-trisphosphate- and nicotinic acid dinucleotide phosphate-sensitive channels and maintained over time by store-operated Ca 2+ entry. In addition, histamine evoked robust NO release that was prevented by interfering with the accompanying intracellular Ca 2+ oscillations, thereby confirming that the endothelial NO synthase is recruited by Ca 2+ spikes also in hCMEC/D3 cells. These data provide the first evidence that histamine evokes NO production from human cerebrovascular endothelial cells through intracellular Ca 2+ oscillations, thereby shedding novel light on the mechanisms by which this neuromodulator controls cerebral blood flow.


Asunto(s)
Encéfalo/irrigación sanguínea , Calcio/metabolismo , Células Endoteliales/efectos de los fármacos , Histamina/farmacología , Microvasos/citología , Óxido Nítrico/metabolismo , Línea Celular , Células Endoteliales/metabolismo , Agonistas de los Receptores Histamínicos/farmacología , Humanos , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , NADP/análogos & derivados , NADP/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo
6.
Sci Rep ; 9(1): 16693, 2019 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-31723148

RESUMEN

Oxygen diffusion across the air-blood barrier in the lung is commensurate with metabolic needs and ideally allows full equilibration between alveolar and blood partial oxygen pressures. We estimated the alveolo-capillary O2 equilibration in 18 healthy subjects at sea level at rest and after exposure to increased O2 demand, including work at sea level and on hypobaric hypoxia exposure at 3840 m (PA ~ 50 mmHg). For each subject we estimated O2 diffusion capacity (DO2), pulmonary capillary blood volume (Vc) and cardiac output ([Formula: see text]). We derived blood capillary transit time [Formula: see text] and the time constant of the equilibration process ([Formula: see text], ß being the slope of the hemoglobin dissociation curve). O2 equilibration at the arterial end of the pulmonary capillary was defined as [Formula: see text]. Leq greately differed among subjects in the most demanding O2 condition (work in hypoxia): lack of full equilibration was found to range from 5 to 42% of the alveolo-capillary PO2 gradient at the venous end. The present analysis proves to be sensible enough to highlight inter-individual differences in alveolo-capillary equilibration among healthy subjects.


Asunto(s)
Barrera Alveolocapilar/fisiopatología , Ejercicio Físico , Hipoxia/fisiopatología , Consumo de Oxígeno , Oxígeno/sangre , Capacidad de Difusión Pulmonar , Adulto , Gasto Cardíaco , Femenino , Voluntarios Sanos , Humanos , Masculino , Ventilación Pulmonar
7.
J Cell Physiol ; 234(4): 4540-4562, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30191989

RESUMEN

Basal forebrain neurons control cerebral blood flow (CBF) by releasing acetylcholine (Ach), which binds to endothelial muscarinic receptors to induce nitric (NO) release and vasodilation in intraparenchymal arterioles. Nevertheless, the mechanism whereby Ach stimulates human brain microvascular endothelial cells to produce NO is still unknown. Herein, we sought to assess whether Ach stimulates NO production in a Ca2+ -dependent manner in hCMEC/D3 cells, a widespread model of human brain microvascular endothelial cells. Ach induced a dose-dependent increase in intracellular Ca2+ concentration ([Ca2+ ]i ) that was prevented by the genetic blockade of M5 muscarinic receptors (M5-mAchRs), which was the only mAchR isoform coupled to phospholipase Cß (PLCß) present in hCMEC/D3 cells. A comprehensive real-time polymerase chain reaction analysis revealed the expression of the transcripts encoding for type 3 inositol-1,4,5-trisphosphate receptors (InsP3 R3), two-pore channels 1 and 2 (TPC1-2), Stim2, Orai1-3. Pharmacological manipulation showed that the Ca2+ response to Ach was mediated by InsP3 R3, TPC1-2, and store-operated Ca2+ entry (SOCE). Ach-induced NO release, in turn, was inhibited in cells deficient of M5-mAchRs. Likewise, Ach failed to increase NO levels in the presence of l-NAME, a selective NOS inhibitor, or BAPTA, a membrane-permeant intracellular Ca2+ buffer. Moreover, the pharmacological blockade of the Ca2+ response to Ach also inhibited the accompanying NO production. These data demonstrate for the first time that synaptically released Ach may trigger NO release in human brain microvascular endothelial cells by stimulating a Ca2+ signal via M5-mAchRs.


Asunto(s)
Acetilcolina/farmacología , Señalización del Calcio/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Microvasos/efectos de los fármacos , Agonistas Muscarínicos/farmacología , Acoplamiento Neurovascular/efectos de los fármacos , Óxido Nítrico/metabolismo , Prosencéfalo/irrigación sanguínea , Receptor Muscarínico M5/agonistas , Canales de Calcio/genética , Canales de Calcio/metabolismo , Canales de Calcio Activados por la Liberación de Calcio/genética , Canales de Calcio Activados por la Liberación de Calcio/metabolismo , Células Cultivadas , Células Endoteliales/metabolismo , Humanos , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Microvasos/metabolismo , Receptor Muscarínico M5/genética , Receptor Muscarínico M5/metabolismo , Molécula de Interacción Estromal 2/genética , Molécula de Interacción Estromal 2/metabolismo , Transmisión Sináptica
8.
J Cell Physiol ; 234(4): 3538-3554, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30451297

RESUMEN

The neurotransmitter glutamate increases cerebral blood flow by activating postsynaptic neurons and presynaptic glial cells within the neurovascular unit. Glutamate does so by causing an increase in intracellular Ca2+ concentration ([Ca2+ ]i ) in the target cells, which activates the Ca2+ /Calmodulin-dependent nitric oxide (NO) synthase to release NO. It is unclear whether brain endothelial cells also sense glutamate through an elevation in [Ca2+ ]i and NO production. The current study assessed whether and how glutamate drives Ca2+ -dependent NO release in bEND5 cells, an established model of brain endothelial cells. We found that glutamate induced a dose-dependent oscillatory increase in [Ca2+ ]i , which was maximally activated at 200 µM and inhibited by α-methyl-4-carboxyphenylglycine, a selective blocker of Group 1 metabotropic glutamate receptors. Glutamate-induced intracellular Ca2+ oscillations were triggered by rhythmic endogenous Ca2+ mobilization and maintained over time by extracellular Ca2+ entry. Pharmacological manipulation revealed that glutamate-induced endogenous Ca2+ release was mediated by InsP3 -sensitive receptors and nicotinic acid adenine dinucleotide phosphate (NAADP) gated two-pore channel 1. Constitutive store-operated Ca2+ entry mediated Ca2+ entry during ongoing Ca2+ oscillations. Finally, glutamate evoked a robust, although delayed increase in NO levels, which was blocked by pharmacologically inhibition of the accompanying intracellular Ca2+ signals. Of note, glutamate induced Ca2+ -dependent NO release also in hCMEC/D3 cells, an established model of human brain microvascular endothelial cells. This investigation demonstrates for the first time that metabotropic glutamate-induced intracellular Ca2+ oscillations and NO release have the potential to impact on neurovascular coupling in the brain.


Asunto(s)
Encéfalo/irrigación sanguínea , Señalización del Calcio/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Ácido Glutámico/farmacología , Inositol 1,4,5-Trifosfato/metabolismo , NADP/análogos & derivados , Acoplamiento Neurovascular/efectos de los fármacos , Óxido Nítrico/metabolismo , Animales , Canales de Calcio/metabolismo , Línea Celular , Relación Dosis-Respuesta a Droga , Células Endoteliales/metabolismo , Humanos , Ratones , NADP/metabolismo , Receptores de Glutamato Metabotrópico/agonistas , Factores de Tiempo
9.
Epilepsia ; 59(9): 1774-1784, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30039519

RESUMEN

OBJECTIVE: The key factors that promote the termination of focal seizures have not been fully clarified. The buildup of neuronal synchronization during seizures has been proposed as one of the possible activity-dependent, self-limiting mechanisms. We investigate if increased thalamo-cortical coupling contributes to enhance synchronization during the late phase of focal seizure-like events (SLEs) generated in limbic regions. METHODS: Recordings were simultaneously performed in the nucleus reuniens of the thalamus, in the hippocampus and in the entorhinal cortex of the isolated guinea pig brain during focal bicuculline-induced SLEs with low voltage fast activity at onset. RESULTS: Spectral coherence and cross-correlation analysis demonstrated a progressive thalamo-cortical entrainment and synchronization in the generation of bursting activity that characterizes the final part of SLEs. The hippocampus is the first activated structure at the beginning of SLE bursting phase and thalamo-hippocampal synchronization is progressively enhanced as SLE develops. The thalamus takes the lead in generating the bursting discharge as SLE end approaches. SIGNIFICANCE: As suggested by clinical studies performed during pre-surgical intracranial monitoring, our data confirm a role of the midline thalamus in leading the synchronous bursting activity at the end of focal seizures in the mesial temporal regions.


Asunto(s)
Hipocampo/fisiopatología , Núcleos Talámicos de la Línea Media/fisiopatología , Vías Nerviosas/fisiopatología , Convulsiones/patología , Potenciales de Acción/fisiología , Análisis de Varianza , Animales , Modelos Animales de Enfermedad , Femenino , Cobayas , Hipocampo/patología , Núcleos Talámicos de la Línea Media/patología , Neuronas/fisiología , Técnicas de Placa-Clamp , Convulsiones/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...